Effect of Alirocumab, a Monoclonal Proprotein Convertase Subtilisin/Kexin 9 Antibody, on...

24
Accepted Manuscript Effect of Alirocumab, a Monoclonal Proprotein Convertase Subtilisin/Kexin 9 Antibody, on Lipoprotein(a) Concentrations (a Pooled Analysis of 150 mg Every 2 Weeks Dosing from Phase 2 Trials) Daniel Gaudet, MD, PhD Dean Kereiakes, MD James McKenney, PharmD Eli Roth, MD Corinne Hanotin, MD Dan Gipe, MD Yunling Du, PhD Anne-Catherine Ferrand, MSc Henry Ginsberg, MD Evan Stein, MD, PhD PII: S0002-9149(14)01299-5 DOI: 10.1016/j.amjcard.2014.05.060 Reference: AJC 20520 To appear in: The American Journal of Cardiology Received Date: 24 March 2014 Revised Date: 15 May 2014 Accepted Date: 28 May 2014 Please cite this article as: Gaudet D, Kereiakes D, McKenney J, Roth E, Hanotin C, Gipe D, Du Y, Ferrand A-C, Ginsberg H, Stein E, Effect of Alirocumab, a Monoclonal Proprotein Convertase Subtilisin/Kexin 9 Antibody, on Lipoprotein(a) Concentrations (a Pooled Analysis of 150 mg Every 2 Weeks Dosing from Phase 2 Trials), The American Journal of Cardiology (2014), doi: 10.1016/ j.amjcard.2014.05.060. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Transcript of Effect of Alirocumab, a Monoclonal Proprotein Convertase Subtilisin/Kexin 9 Antibody, on...

Accepted Manuscript

Effect of Alirocumab, a Monoclonal Proprotein Convertase Subtilisin/Kexin 9Antibody, on Lipoprotein(a) Concentrations (a Pooled Analysis of 150 mg Every 2Weeks Dosing from Phase 2 Trials)

Daniel Gaudet, MD, PhD Dean Kereiakes, MD James McKenney, PharmD Eli Roth,MD Corinne Hanotin, MD Dan Gipe, MD Yunling Du, PhD Anne-Catherine Ferrand,MSc Henry Ginsberg, MD Evan Stein, MD, PhD

PII: S0002-9149(14)01299-5

DOI: 10.1016/j.amjcard.2014.05.060

Reference: AJC 20520

To appear in: The American Journal of Cardiology

Received Date: 24 March 2014

Revised Date: 15 May 2014

Accepted Date: 28 May 2014

Please cite this article as: Gaudet D, Kereiakes D, McKenney J, Roth E, Hanotin C, Gipe D, DuY, Ferrand A-C, Ginsberg H, Stein E, Effect of Alirocumab, a Monoclonal Proprotein ConvertaseSubtilisin/Kexin 9 Antibody, on Lipoprotein(a) Concentrations (a Pooled Analysis of 150 mg Every2 Weeks Dosing from Phase 2 Trials), The American Journal of Cardiology (2014), doi: 10.1016/j.amjcard.2014.05.060.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service toour customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, and alllegal disclaimers that apply to the journal pertain.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

1

Effect of Alirocumab, a Monoclonal Proprotein Convertase

Subtilisin/Kexin 9 Antibody, on Lipoprotein(a)

Concentrations (a Pooled Analysis of 150 mg Every 2

Weeks Dosing from Phase 2 Trials)

Daniel Gaudet, MD, PhDa,*, Dean Kereiakes, MDb, James McKenney, PharmDc, Eli

Roth, MDd, Corinne Hanotin, MDe, Dan Gipe, MDf, Yunling Du, PhDf, Anne-

Catherine Ferrand, MSce, Henry Ginsberg, MDg, Evan Stein, MD, PhDh

aThe ECOGENE-21 Clinical Research Center and Lipid Clinic, Department of

Medicine, Université de Montreal, Chicoutimi, Quebec, Canada; bThe Christ Hospital

Heart and Vascular Center/The Lindner Research Center, Cincinnati, OH, USA;

cVirginia Commonwealth University, Richmond, VA, USA; dSterling Research Group,

Cincinnati, OH, USA; eSanofi, Paris, France; fRegeneron Pharmaceuticals, Inc.,

Tarrytown, NY, USA; gColumbia University, New York, NY, USA; hMetabolic and

Atherosclerotic Research Center, Cincinnati, OH, USA

Running title: Alirocumab Reduces Plasma Lipoprotein(a) Levels

Sources of funding: This analysis was funded by Sanofi and Regeneron.

*Corresponding author: Dr. Daniel Gaudet, PhD, Department of Medicine,

Université de Montréal, ECOGENE 21 Clinical Research Center, Chicoutimi

Hospital, Chicoutimi, G7H 5H6, Canada, Tel: 418-541-1077; Fax: 418-541-1116;

E-mail address: [email protected]

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

2

Abstract

Lipoprotein(a) [Lp(a)] is an independent risk factor for cardiovascular disease, with

limited treatment options. This analysis evaluated the effect of a monoclonal

antibody to proprotein convertase subtilisin/kexin 9 (PCSK9), alirocumab 150 mg

every 2 weeks (Q2W), on Lp(a) levels in pooled data from 3 double-blind,

randomized, placebo-controlled phase 2 studies of 8 or 12 weeks duration

conducted in patients with hypercholesterolemia on background lipid-lowering

therapy (NCT01266876, NCT01288469, NCT01288443). Data were available for

102/108 patients who received alirocumab 150 mg Q2W and 74/77 patients who

received placebo. Alirocumab resulted in a significant reduction in Lp(a) from

baseline compared with placebo (–30.3% versus –0.3%, p <0.0001). Median

percentage Lp(a) reductions in the alirocumab group were of a similar magnitude

across a range of baseline Lp(a) levels, resulting in greater absolute reductions in

Lp(a) in patients with higher baseline levels. Regression analysis indicated that less

than 5% of the variance in the reduction of Lp(a) was explained by the effect of

alirocumab on low-density lipoprotein cholesterol (LDL-C). In conclusion, pooled data

from 3 phase 2 trials demonstrate substantive reduction in Lp(a) with alirocumab 150

mg Q2W, including patients with baseline Lp(a) >50 mg/dl. Reductions in Lp(a) only

weakly correlated with the magnitude of LDL-C lowering.

Keywords: Lipoprotein(a); low-density lipoprotein cholesterol; alirocumab;

monoclonal antibodies; PCSK9

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

3

Introduction

Elevated lipoprotein(a) [Lp(a)] levels are considered to be an independent risk

factor for cardiovascular disease, with risk continuing to increase as levels rise.1,2

The 2011 European Atherosclerosis Society (EAS)/European Society of Cardiology

dyslipidemia management guidelines recommend measurement of Lp(a) in high-risk

patients or in those with a family history of premature cardiovascular disease,3 and

the EAS define high-risk Lp(a) levels as >50 mg/dl, approximately the 80th percentile

in the Caucasian population.2 Of the widely prescribed lipid-modifying therapies, only

nicotinic acid consistently reduces Lp(a).2 Despite the potent effect of statins on LDL-

C levels, in general they show little to no effect on Lp(a) in clinical trials.4 Alirocumab

(formerly SAR236553/REGN727), a highly specific, fully human monoclonal antibody

to proprotein convertase subtilisin/kexin type 9 (PCSK9), significantly (p <0.001)

reduced LDL cholesterol (LDL-C) by up to 72.4% when combined with statin or other

lipid-lowering therapy in phase 2 trials, and showed a trend to reduce Lp(a), a

secondary parameter, across all dosing regimens tested.5–7 The objective of the

present pooled analysis of alirocumab 150 mg Q2W, a common dose in all phase 2

trials, was to provide, post-hoc, a more robust assessment of its effects on Lp(a),

and examine the relationship with baseline levels and LDL-C reductions.

Methods

Data were pooled from 3 double-blind, randomized, placebo-controlled phase 2

studies with alirocumab of 8- or 12-weeks’ duration (NCT01288443; NCT01266876;

NCT01288469). The study designs (Supplementary Figure 1) have been previously

described.5–7 Briefly, in these studies a total of 352 patients with heterozygous

familial hypercholesterolemia or non-familial forms of hypercholesterolemia and LDL-

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

4

C ≥100 mg/dl under background statin or statin plus ezetimibe 10 mg treatment were

randomized to alirocumab 50–300 mg Q2W or every 4 weeks (n = 275) or placebo

(n = 77). One of the studies included up-titration of background atorvastatin from 10

mg to 80 mg in the placebo arm and 1 of 2 alirocumab arms; in the other alirocumab

arm patients remained at a background atorvastatin dose of 10 mg.7 This study was

included in the pooled analysis since impact of statin uptitration on Lp(a) was

expected to be low based on previous studies.4 This analysis focuses on the dose

regimen alirocumab 150 mg Q2W, which was common to all 3 phase 2 studies and

was considered to provide consistent, robust effects on LDL-C;5,6 this is also one of

the doses being evaluated in the alirocumab phase 3 clinical trials.

In all phase 2 studies, Lp(a) levels were measured at the same laboratory Lp(a)

using rate immunonephelometry (Dade Behring BNII nephelometer, Siemens

Healthcare Diagnostics, Deerfield, Illinois).5,6 Data on Lp(a) levels at baseline and

end of treatment (week 8/12 on-treatment value or the last available on-treatment

value carried forward) from the modified intention-to-treat populations of the

3 studies were pooled, and percentage changes from baseline for alirocumab

150 mg Q2W and placebo were compared using analysis of covariance with

treatment group and study as fixed effects and baseline Lp(a) as covariate. P-values

associated with these exploratory analyses are provided for descriptive purposes

only and were not adjusted for multiplicity. The relationship between the percentage

changes from baseline in Lp(a) and LDL-C was assessed using linear regression,

and the Spearman’s correlation coefficient was calculated.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

5

Results

Baseline characteristics (Table 1) were well-balanced. All patients were

receiving background statin therapy and 11/108 patients (10%) in the alirocumab

group and 11/77 patients (14%) in the placebo group also received ezetimibe 10 mg.

Baseline and on-treatment Lp(a) data were available for 102 of the 108 patients who

received alirocumab and 74 of the 77 patients who received placebo (Table 2).

Thirty-six (34%) patients treated with alirocumab and 25 (33%) patients treated with

placebo had baseline Lp(a) >50 mg/dl, the high-risk cut-point by the EAS guidelines.2

Absolute and percentage median reductions from baseline in Lp(a) are shown

in Figure 1A and 1B, respectively. The median absolute reductions in Lp(a) from

baseline were substantially greater in patients with the higher baseline Lp(a) (Figure

1A). Reductions in Lp(a) were only weakly correlated with the magnitude of LDL-C

lowering (Spearman’s correlation coefficient: 0.2236; Supplementary Figure 2A).

Similar results were observed using a regression analysis comparing actual

achieved LDL-C levels and percentage reduction in Lp(a) (Supplementary

Figure 2B). The percentage reductions in Lp(a) from baseline with alirocumab were

consistent between a pool of the 2 12-week studies and a pool of the two arms

receiving alirocumab in the 8-week study (–28.3% and –32.1%, respectively).

The most common treatment-emergent adverse event in the alirocumab phase

2 trials was injection site reaction, typically episodic and of mild intensity and short

duration; five serious adverse events occurred in 4 patients (1.5%) who received

alirocumab.5–7

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

6

Discussion

This analysis of pooled data from the phase 2 studies with alirocumab 150 mg

Q2W compared with placebo demonstrated robust and statistically significant

(p <0.0001) reductions in Lp(a) of ~30%. This compares favorably with those

reported with extended-release nicotinic acid 2 g/day (~20–35%),8,9 and is consistent

with those reported with the PCSK9 monoclonal antibody evolocumab (18–32%

versus placebo).10,11 As reported for evolocumab, the absolute (mg/dl) reductions in

Lp(a) with alirocumab showed a greater reduction in the patients considered by

treatment guidelines to have elevated Lp(a) levels.2,10,11

The effect of statins on Lp(a) has been extensively evaluated over the past

3 decades and in all large well-controlled studies there has been no or only minimal

effects.4,12 Of the widely prescribed lipid-lowering therapies, only nicotinic acid 1–3 g

per day has been shown to consistently reduce Lp(a) levels.2 Recently, reductions in

Lp(a) of 21–33% have been reported with apo B synthesis inhibition using

mipomersen,13,14 and of 15–17% with microsomal triglyceride transfer protein

inhibition using lomitapide.15 However, these drugs are approved only for rare

patients with homozygous familial hypercholesterolemia. Inhibition of cholesteryl

ester transfer protein (CETP) with anacetrapib was reported to reduce Lp(a) by 17–

24%,16 however this drug is still in development and other CETP-inihibitors

(torcetrapib, dalcetrapib) have so far failed to demonstrate a positive effect on

cardiovascular events and be approved for use. In addition to drug therapy,

apheresis reduces high Lp(a) levels, but its use in routine practice is also limited.2

The mechanisms by which Lp(a) is synthesized, metabolized and cleared from

the circulation are poorly understood (Figure 2), and the mechanism by which

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

7

PCSK9 monoclonal antibodies reduce Lp(a) is also unclear. Both statins and

alirocumab are thought to reduce LDL-C and most other apo B-containing

lipoproteins by increasing the number of LDL receptors on hepatocytes: statins via

HMG-CoA inhibition,17 alirocumab via PCSK9 blockade.18 It is generally considered

that the Lp(a) particle has poor affinity for the LDL receptor.19 One hypothesis is that

the further upregulation of the LDL receptor combined with the very low LDL and apo

B levels achieved with alirocumab treatment allows for uptake of Lp(a) by LDL

receptors. For example, the mean achieved LDL-C level for the 150 mg Q2W arms

in these 3 studies ranged from 34.2 mg/dl to 50.3 mg/dl, mean levels that are lower

than those typically achieved in randomized trials of statins alone (~60–100

mg/dl).20,21

Regression analysis conducted in this study (Supplementary Figure 2) showed

that a portion of the observed reduction in Lp(a) was not dependent on the

magnitude of LDL-C lowering or LDL-C level achieved. In addition the JUPITER trial

achieved LDL-C levels <50 mg/dl in 7,746 patients with no reductions seen in

Lp(a).22 These data suggest an effect of alirocumab on Lp(a) production or fractional

clearance that may be independent of, or in addition to, the mechanism of LDL-C

lowering. The LDL receptor independent reduction is supported by the recent report

of a 15–20% decrease in Lp(a) in 2 LDL receptor-negative homozygous familial

hypercholesterolemia patients treated with evolocumab, who had no associated

reduction in LDL-C.23 However, two reports on the effects of evolocumab on

Lp(a)10,11 indicated a stronger correlation between Lp(a) and LDL-C reductions than

what was observed in the present analysis; this discrepancy may be due to the

larger patient numbers increasing the statistical power of the evolocumab studies.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

8

In terms of synthesis, apo(a), one of the principal components of Lp(a), is

expressed by hepatocytes and released into the circulation where the assembly of

apo(a) and LDL apo B-containing particles bind to form Lp(a) particles at the outer

hepatocyte surface.24 The ability to reduce Lp(a) by either inhibition of LDL-apo B or

apo(a) synthesis indicates the requirement of both for its formation.14,25 The liver

contributes to Lp(a) removal from the circulation26 although, as mentioned above, the

mechanism is unclear.19 The kidney as well as peripheral tissues may also play an

important role in Lp(a) clearance.27 New, emerging receptors have also been

proposed, although their respective contributions to Lp(a) clearance remain to be

determined. Such receptors include, among others, sorting receptors (such as

sortilin28), endocytic receptors (such as the syndecan-1 heparan sulfate

proteoglycan29) or docking receptors. The effect of PCSK9 inhibition on the

regulation of receptors other than the LDL receptor is still under investigation.

The present analysis did not take into consideration variations in the number of

kringle IV repeats in the LPA gene encoding apo(a), which account for ~25–30% of

the variation in Lp(a) plasma concentrations.2,30 However, we did not observe a

relationship between Lp(a) concentration and the response to alirocumab in terms of

LDL-C lowering. Approximately 30% of patients in this analysis had Lp(a) >50 mg/dl

at baseline. High Lp(a) levels are known to affect LDL-C levels calculated using the

Friedewald method, with ~8% of the cholesterol measured as LDL-C estimated to be

due to Lp(a) in patients with baseline Lp(a) 30–60 mg/dl, and a corresponding figure

of ~20% with Lp(a) >60 mg/dl.4 This was not taken into account for the regression

analysis; to correct for the cholesterol in LDL-C which was due to Lp(a), previous

studies have subtracted the Lp(a) mass, multiplied by 0.3, from the LDL-C values.4

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

9

Acknowledgments

Medical writing support was provided by Rob Campbell of Prime Medica Ltd,

Knutsford, Cheshire, UK, supported by Sanofi and Regeneron.

Disclosures

D. Gaudet has worked as a consultant and on advisory boards for Sanofi,

Regeneron, Novartis, Isis, Catabasis, Aegerion, and Amgen (all modest).

D. Kereiakes has no relevant financial disclosures. J.M. McKenney and E. Roth are

employed by companies that have received research funds and have received

consulting fees from Regeneron and/or Sanofi (significant). D. Gipe and Y. Du are

employees of Regeneron (both significant). C. Hanotin and A.-C. Ferrand are

employees of Sanofi (both significant). H. Ginsberg has received research funding

from Sanofi (significant) and has worked as a consultant and on advisory boards for

Sanofi and Regeneron (modest). E. Stein has received consultant fees from Sanofi,

Regeneron, Amgen, BMS/Adnexus, and Roche/Genentech (all modest).

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

10

1. Nicholls SJ, Tang WH, Scoffone H, Brennan DM, Hartiala J, Allayee H, Hazen

SL. Lipoprotein(a) levels and long-term cardiovascular risk in the contemporary

era of statin therapy. J Lipid Res 2010;51:3055–3061.

2. Nordestgaard BG, Chapman MJ, Ray K, Boren J, Andreotti F, Watts GF,

Ginsberg H, Amarenco P, Catapano A, Descamps OS, Fisher E, Kovanen PT,

Kuivenhoven JA, Lesnik P, Masana L, Reiner Z, Taskinen MR, Tokgozoglu L,

Tybjaerg-Hansen A. Lipoprotein(a) as a cardiovascular risk factor: current status.

Eur Heart J 2010;31:2844–2853.

3. Reiner Z, Catapano AL, De BG, Graham I, Taskinen MR, Wiklund O, Agewall S,

Alegria E, Chapman MJ, Durrington P, Erdine S, Halcox J, Hobbs R, Kjekshus J,

Filardi PP, Riccardi G, Storey RF, Wood D. ESC/EAS Guidelines for the

management of dyslipidaemias: the Task Force for the management of

dyslipidaemias of the European Society of Cardiology (ESC) and the European

Atherosclerosis Society (EAS). Eur Heart J 2011;32:1769–1818.

4. Deshmukh HA, Colhoun HM, Johnson T, McKeigue PM, Betteridge DJ,

Durrington PN, Fuller JH, Livingstone S, Charlton-Menys V, Neil A, Poulter N,

Sever P, Shields DC, Stanton AV, Chatterjee A, Hyde C, Calle RA, Demicco DA,

Trompet S, Postmus I, Ford I, Jukema JW, Caulfield M, Hitman GA. Genome-

wide association study of genetic determinants of LDL-c response to atorvastatin

therapy: importance of Lp(a). J Lipid Res 2012;53:1000–1011.

5. Stein EA, Gipe D, Bergeron J, Gaudet D, Weiss R, Dufour R, Wu R, Pordy R.

Effect of a monoclonal antibody to PCSK9, REGN727/SAR236553, to reduce

low-density lipoprotein cholesterol in patients with heterozygous familial

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

11

hypercholesterolaemia on stable statin dose with or without ezetimibe therapy: a

phase 2 randomised controlled trial. Lancet 2012;380:29–36.

6. McKenney JM, Koren MJ, Kereiakes DJ, Hanotin C, Ferrand AC, Stein EA.

Safety and efficacy of a monoclonal antibody to proprotein convertase

subtilisin/kexin type 9 serine protease, SAR236553/REGN727, in patients with

primary hypercholesterolemia receiving ongoing stable atorvastatin therapy. J Am

Coll Cardiol 2012;59:2344–2353.

7. Roth EM, McKenney JM, Hanotin C, Asset G, Stein EA. Atorvastatin with or

without an antibody to PCSK9 in primary hypercholesterolemia. N Engl J Med

2012;367:1891–1900.

8. Guyton JR, Blazing MA, Hagar J, Kashyap ML, Knopp RH, McKenney JM, Nash

DT, Nash SD. Extended-release niacin vs gemfibrozil for the treatment of low

levels of high-density lipoprotein cholesterol. Niaspan-Gemfibrozil Study Group.

Arch Intern Med 2000;160:1177–1184.

9. Maccubbin D, Bays HE, Olsson AG, Elinoff V, Elis A, Mitchel Y, Sirah W,

Betteridge A, Reyes R, Yu Q, Kuznetsova O, Sisk CM, Pasternak RC, Paolini JF.

Lipid-modifying efficacy and tolerability of extended-release niacin/laropiprant in

patients with primary hypercholesterolaemia or mixed dyslipidaemia. Int J Clin

Pract 2008;62:1959–1970.

10. Desai NR, Kohli P, Giugliano RP, O'Donoghue ML, Somaratne R, Zhou J,

Hoffman EB, Huang F, Rogers WJ, Wasserman SM, Scott R, Sabatine MS.

AMG145, a monoclonal antibody against proprotein convertase subtilisin kexin

type 9, significantly reduces lipoprotein(a) in hypercholesterolemic patients

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

12

receiving statin therapy: an analysis from the LDL-C Assessment with Proprotein

Convertase Subtilisin Kexin Type 9 Monoclonal Antibody Inhibition Combined

with Statin Therapy (LAPLACE)-Thrombolysis in Myocardial Infarction (TIMI) 57

trial. Circulation 2013;128:962–969.

11. Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Langslet G, Bays H, Blom D,

Eriksson M, Dent R, Wasserman SM, Huang F, Xue A, Albizem M, Scott R, Stein

EA. Reduction in lipoprotein (a) with the PCSK9 monoclonal antibody

evolocumab (AMG 145): a pooled analysis of over 1300 patients in 4 phase 2

trials. J Am Coll Cardiol 2014;Jan 27. pii: S0735-1097(14)00281-2. [Epub ahead

of print]

12. Hunninghake DB, Stein EA, Mellies MJ. Effects of one year of treatment with

pravastatin, an HMG-CoA reductase inhibitor, on lipoprotein a. J Clin Pharmacol

1993;33:574–580.

13. McGowan MP, Tardif JC, Ceska R, Burgess LJ, Soran H, Gouni-Berthold I,

Wagener G, Chasan-Taber S. Randomized, placebo-controlled trial of

mipomersen in patients with severe hypercholesterolemia receiving maximally

tolerated lipid-lowering therapy. PLoS One 2012;7:e49006

14. Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, Chin W, Tribble

DL, McGowan M. Apolipoprotein B synthesis inhibition with mipomersen in

heterozygous familial hypercholesterolemia: results of a randomized, double-

blind, placebo-controlled trial to assess efficacy and safety as add-on therapy in

patients with coronary artery disease. Circulation 2012;126:2283–2292.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

13

15. Cuchel M, Meagher EA, du Toit TH, Blom DJ, Marais AD, Hegele RA, Averna

MR, Sirtori CR, Shah PK, Gaudet D, Stefanutti C, Vigna GB, Du Plessis AM,

Propert KJ, Sasiela WJ, Bloedon LT, Rader DJ. Efficacy and safety of a

microsomal triglyceride transfer protein inhibitor in patients with homozygous

familial hypercholesterolaemia: a single-arm, open-label, phase 3 study. Lancet

2013;381:40–46.

16. Cannon CP, Shah S, Dansky HM, Davidson M, Brinton EA, Gotto AM,

Stepanavage M, Liu SX, Gibbons P, Ashraf TB, Zafarino J, Mitchel Y, Barter P.

Safety of anacetrapib in patients with or at high risk for coronary heart disease. N

Engl J Med 2010;363:2406–2415.

17. Goldstein JL, Brown MS. The LDL receptor. Arterioscler Thromb Vasc Biol

2009;29:431–438.

18. Stein EA, Raal FJ. Insights into PCSK9, low-density lipoprotein receptor, and

low-density lipoprotein cholesterol metabolism: of mice and man. Circulation

2013;127:2372–2374.

19. Rader DJ, Mann WA, Cain W, Kraft HG, Usher D, Zech LA, Hoeg JM,

Davignon J, Lupien P, Grossman M, . The low density lipoprotein receptor is not

required for normal catabolism of Lp(a) in humans. J Clin Invest 1995;95:1403–

1408.

20. Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, Joyal

SV, Hill KA, Pfeffer MA, Skene AM. Intensive versus moderate lipid lowering with

statins after acute coronary syndromes. N Engl J Med 2004;350:1495–1504.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

14

21. Colhoun HM, Betteridge DJ, Durrington PN, Hitman GA, Neil HA, Livingstone

SJ, Thomason MJ, Mackness MI, Charlton-Menys V, Fuller JH. Primary

prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the

Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised

placebo-controlled trial. Lancet 2004;364:685–696.

22. Khera AV, Everett BM, Caulfield MP, Hantash FM, Wohlgemuth J, Ridker PM,

Mora S. Lipoprotein(a) concentrations, rosuvastatin therapy, and residual

vascular risk: an analysis from the JUPITER Trial (justification for the use of

statins in prevention: an intervention trial valuating rosuvastatin). Circulation

2013;129:635–642.

23. Stein EA, Honarpour N, Wasserman SM, Xu F, Scott R, Raal FJ. Effect of the

proprotein convertase subtilisin/kexin 9 monoclonal antibody, AMG 145, in

homozygous familial hypercholesterolemia. Circulation 2013;128:2113–2120.

24. White AL, Lanford RE. Cell surface assembly of lipoprotein(a) in primary

cultures of baboon hepatocytes. J Biol Chem 1994;269:28716–28723.

25. Viney N, Graham M, Crooke R, Hughes S, Singleton W. Evaluation of ISIS

apo(a)Rx, an antisense inhibitor to apolipoprotein(a), in healthy volunteers.

Circulation 2013;128Abstract A14196.

26. Cain WJ, Millar JS, Himebauch AS, Tietge UJ, Maugeais C, Usher D, Rader

DJ. Lipoprotein [a] is cleared from the plasma primarily by the liver in a process

mediated by apolipoprotein [a]. J Lipid Res 2005;46:2681–2691.

27. Albers JJ, Koschinsky ML, Marcovina SM. Evidence mounts for a role of the

kidney in lipoprotein(a) catabolism. Kidney Int 2007;71:961–962.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

15

28. Strong A, Ding Q, Edmondson AC, Millar JS, Sachs KV, Li X, Kumaravel A,

Wang MY, Ai D, Guo L, Alexander ET, Nguyen D, Lund-Katz S, Phillips MC,

Morales CR, Tall AR, Kathiresan S, Fisher EA, Musunuru K, Rader DJ. Hepatic

sortilin regulates both apolipoprotein B secretion and LDL catabolism. J Clin

Invest 2012;122:2807–2816.

29. van Barlingen HH, Kleinveld HA, Erkelens DW, de Bruin TW. Lipoprotein

lipase-enhanced binding of lipoprotein(a) [Lp(a)] to heparan sulfate is improved

by apolipoprotein E (apoE) saturation: secretion-capture process of apoE is a

possible route for the catabolism of Lp(a). Metabolism 1997;46:650–655.

30. Lanktree MB, Anand SS, Yusuf S, Hegele RA. Comprehensive analysis of

genomic variation in the LPA locus and its relationship to plasma lipoprotein(a) in

South Asians, Chinese, and European Caucasians. Circ Cardiovasc Genet

2010;3:39–46.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

16

Figure 1. Effect of alirocumab 150 mg Q2W on Lp(a) (pooled modified intention-to-

treat population). Shown are absolute changes from baseline (A) or percentage

changes from baseline (B) in Lp(a), with patients subdivided by baseline Lp(a)

levels.

*p <0.0001 versus placebo.

P-values were derived from analysis of covariance with treatment group and study as fixed effects and

baseline Lp(a) as covariate and are provided for descriptive purposes only, and were not adjusted for multiplicity.

IQR = inter-quartile range; LOCF = last observation carried forward; Lp(a) = lipoprotein(a); Q2W = every

2 weeks; SD = standard deviation.

Figure 2. Biology of Lp(a).

(1) Lp(a) consists in an apo(a) covalently bound to the apo B-100 component of a LDL-like particle. The

Lp(a) proteic backbone [apo(a) and apoB-100] is synthesized in the liver, whereas the Lp(a) assembly is

suspected to take place at the outer hepatocyte surface. Apo(a) proteins vary in size due to a variable number of

kringle IV repeats in the LPA gene. (2) The half-life of Lp(a) in circulation is >3 days. Although the mechanisms

through which Lp(a) promotes atherosclerosis and atherothrombosis are not clearly understood, proposed

mechanisms include inflammatory cell recruitment, increased Lp(a)-cholesterol entrapment in the intima,

transport of proatherogenic oxidized phospholipids and platelet activation, impairing fibrinolysis by inhibition of

plasminogen activation, and enhancing coagulation by inhibition of the tissue factor pathway inhibitor. (3) Lp(a)

has been recently associated with valvular calcification and aortic stenosis. (4) The pathways of Lp(a) clearance

are unclear. The liver seems to be importantly involved, although the contribution of the LDL-receptor in this

process appears to be less important than expected. Several clearance receptors have been identified, although

their respective contributions to Lp(a) removal remain to be determined. (5) The kidney appears to contribute to

Lp(a) clearance. Peripheral tissues also may contribute to Lp(a) removal from the plasma.

Apo = apolipoprotein; LDL, low-density lipoprotein; Lp(a), lipoprotein(a); LPA = apolipoprotein(a) gene;

PAI-1 = plasminogen activator inhibitor-1; TFPI = tissue factor pathway inhibitor; SMC = smooth muscle cells.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Table 1 Patient baseline characteristics (pooled randomized population)

Mean ± standard deviation unless otherwise stated

Placebo (n = 77)

Alirocumab 150 mg every 2 weeks (n = 108)

Age (years) 54 ± 9 58 ± 10

Males 38 (49%) 47 (44%)

White or European American 65 (84%) 96 (89%)

Black or African American 10 (13%) 12 (11%)

Other race 2 (3%) 0

Body mass index (kilograms per meter2)

29 ± 5 29 ± 5

Low-density lipoprotein cholesterol (milligrams per deciliter)

131 ± 28 127 ±25

Total cholesterol (milligrams per deciliter)

211 ± 32 208 ± 31

High-density lipoprotein cholesterol (milligrams per deciliter)

52 ± 14 54 ± 15

Non-high-density lipoprotein cholesterol (milligrams per deciliter)

159 ± 31 154 ± 31

Triglycerides, median (interquartile range) (milligrams per deciliter)

123 (92–174) 124 (88–169)

Apolipoprotein B* (milligrams per deciliter)

109 ± 23 108 ± 24

*n = 76 and n = 107 patients randomized to placebo and alirocumab 150 mg Q2W, respectively,

had apolipoprotein B data available at baseline.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Table 2

Baseline lipoprotein(a) levels in patients included in the pooled analysis

(pooled modified-intent-to-treat population*)

________Variable_____________ Placebo (n = 74)

Alirocumab 150 mg every 2 weeks

(n = 102)_______

Lipoprotein(a), median (interquartile range) (milligrams per deciliter)

19 (6–77) 30 (8–70)

Range, milligrams per deciliter 2–299 2–181

Patients subdivided by baseline lipoprotein(a)

≤50 mg/dl 49 (66%) 68 (67%)

>50 mg/dl 25 (34%) 36 (35%)

*Patients with Lp(a) data available at baseline and end of treatment (week 8/12 on-treatment

value or the last available on-treatment value carried forward).

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Supplementary Figure 1. Summary of design and dosing in the phase 2

studies included in this analysis.

Study 11566 included up-titration of ATV 10 mg to 80 mg in the placebo arm and 1 of the

alirocumab arms.

†Numbers indicate randomized population.

ATV = atorvastatin; FH = familial hypercholesterolemia; HC = hypercholesterolemia;

HeFH = heterozygous familial hypercholesterolemia; Q2W = every 2 weeks; Q4W = every 4 weeks.

MANUSCRIP

T

ACCEPTED

ACCEPTED MANUSCRIPT

Supplementary Figure 2. Relationship between effects of alirocumab 150 mg

Q2W on change from baseline in Lp(a) and either the change from baseline in

LDL-C at end of study treatment (A) or achieved LDL-C (B) (pooled modified

intention-to-treat population).

LDL-C = low-density lipoprotein cholesterol; Lp(a) = lipoprotein(a); Q2W, every 2 weeks.